EP2536428B1 - Vaccins universels contre la grippe à pseudo-particules virales (vlp) - Google Patents

Vaccins universels contre la grippe à pseudo-particules virales (vlp) Download PDF

Info

Publication number
EP2536428B1
EP2536428B1 EP11744998.3A EP11744998A EP2536428B1 EP 2536428 B1 EP2536428 B1 EP 2536428B1 EP 11744998 A EP11744998 A EP 11744998A EP 2536428 B1 EP2536428 B1 EP 2536428B1
Authority
EP
European Patent Office
Prior art keywords
virus
cell
influenza
cells
proteins
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP11744998.3A
Other languages
German (de)
English (en)
Other versions
EP2536428A1 (fr
EP2536428A4 (fr
Inventor
Jose Galarza
George R. Martin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Technovax Inc
Original Assignee
Technovax Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Technovax Inc filed Critical Technovax Inc
Publication of EP2536428A1 publication Critical patent/EP2536428A1/fr
Publication of EP2536428A4 publication Critical patent/EP2536428A4/fr
Application granted granted Critical
Publication of EP2536428B1 publication Critical patent/EP2536428B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16023Virus like particles [VLP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • VLPs Virus-like particles comprising antigenic influenza proteins are described, as are methods for making and using these VLPs.
  • Influenza viruses are the causative agents of respiratory infections in humans and other mammalian and avian species. Unique biological and epidemiological characteristics in the virus life cycle drive a rapid antigenic evolution and continuous emergence of new virus strains, particularly of the type A virus. This continuous emergence of antigenic variants allows for seasonal influenza epidemics and at irregular intervals global pandemics. Annual influenza epidemics are associated with approximately 36,000 deaths in the United States alone and more than 250,000 fatalities worldwide each year while causing enormous health and economical burdens. Pandemic influenza poses a serious threat to human health and such global events as the 1918 pandemic caused an estimated 50 millions death and devastating socio-economical effects.
  • Influenza viruses have a single stranded, segmented RNA genome and are members of the Orthomyxoviridae family. Influenza A viruses display two major glycoproteins on the surface of the virion particle, the hemagglutinin (HA) and the neuraminidase (NA) and based on the antigenic properties of these two molecules they are classified into 16 HA subtypes (H1-H16) and 9 NA subtypes (N1-N9). The predominant immune response following influenza infection targets the surface glycoprotein HA and NA. Antibodies against the HA are able to block virus binding to the cell surface receptor and prevent virus entry and infection. Over time, however, this response is rendered ineffective and unable to protect against emerging antigenic variants resulting from antigenic drift (accumulation of mutations) or antigenic shift (swapping segment by reassortment).
  • HA hemagglutinin
  • NA neuraminidase
  • Vaccine effectiveness depends upon the degree of antigenic similarity between the vaccine and virus strains circulating in humans. Furthermore, a certain proportion of vaccinated individuals, including the elderly, fail to develop protective immunity with the current vaccines.
  • a recent study performed with a phage-display antibody library has identified broadly neutralizing antibodies which are able to block infection across a broad spectrum of influenza virus subtypes ( Jianhua Sui et. al. (2009) Nat. Structural & Mol. Biol. 16:265-273 .)
  • the ability of these antibodies to neutralize a broad spectrum of virus results from reacting with a sub-dominant and highly conserved epitope present in the stem region of the HA molecule, blocking infection by inhibiting membrane fusion rather than preventing receptor binding which is the predominant neutralizing mechanism induced by virus infection or vaccination.
  • Design of a vaccine that displays these subdominant and highly conserved epitopes will elicit broadly neutralizing immune responses able to protect against a broad spectrum of influenza subtypes.
  • viruses are grown by infecting embryonated chicken eggs or cultured cells. This process depends upon the virus's ability to attach via the HA molecule to the sialic acid containing receptor on the cell surface, penetration, virus replication and isolation of viral progeny from the fluids in the egg or cultured cells. Truncation, re-engineering or remodeling of the viral HA would eliminate receptor binding and preclude virus replication and the production of such a vaccine.
  • Influenza VLPs are produced by the simultaneous expression of 2, 3 or 4 viral genes (M1, M2, HA and NA) with M1, M2, or (analogous matrix proteins) HA and NA being the main and essential components of the structure. See, e.g., U.S. Patent Publication Nos. 20080031895 and 20090022762 .
  • the DNA sequences encoding these genes are simultaneously or sequentially transfected into cells, where they are transcribed and translated into their respective proteins that self-assemble into virus-like particles.
  • modifications of the regions of the HA used for receptor binding and virus penetration do not interfere with the production, assembly or release of VLPs from the cells.
  • VLP production allows for the deletion and remodeling of major portions of HA bearing immunodominant and genetically variable regions of the molecule whereas the virus replication method does not allow for these modifications. Similar modification can also be introduced on the NA molecules, which is also displayed on the surface of the VLP vaccine.
  • Kang et al. 2009, Current Topics in Microbiology and Immunology, 333, 269 ) disclose influenza virus-like particles as possible candidates for the preparation of vaccines against influenza. It also describes some examples of virus-like particles, in which the hemagglutinin protein was modified to remove the polybasic cleavage site at the HA1/HA2 junction to reduce pathogenicity. US 2007/184526 and Webby et al.
  • influenza vaccine compositions and methods of making and using such compositions.
  • VLPs virus-like particles
  • compositions comprising these VLPs, as well as methods for making and using these VLPs.
  • the VLPs described herein are devoid of viral genetic material and therefore unable to replicate or cause infection; however given their morphological, biochemical and antigenic similarities to wild type virions, VLPs are highly immunogenic and able to elicit robust protective immune responses.
  • VLPs are not infectious eliminating the need for chemical treatment, thus maintaining the native conformation.
  • the present invention overcomes the issues associated with reformulating influenza vaccines each season, by providing broad protection against current and evolving influenza viruses.
  • VLP virus-like particle
  • at least one matrix protein e.g., an influenza matrix protein such as M1, a thogoto matrix protein and an RSV matrix protein
  • a modified influenza hemagglutinin polypeptide comprising a hemagglutinin1 (HA1) fragment and a hemagglutinin2 (HA2) fragment operably linked to each other, wherein the modification comprises a deletion of at least 150 amino acid residues of a hemagglutinin1 polypeptide, wherein the hemagglutinin2 fragment comprises the transmembrane and cytoplasmic tail domain of the hemagglutinin polypeptide.
  • the modification comprises mutations in one or more amino acids of a hemagglutinin2 polypeptide.
  • the deletion comprises a deletion at least 150 consecutive amino acid residues.
  • the deletion comprises more than one non-consecutive deletions.
  • the influenza polypeptide e.g., HA
  • the influenza polypeptide comprises one or more amino acid mutations (substitutions and/or additions), for example, embodiments in which at least one proteolytic cleavage site and/or at least one linker is inserted into the modified influenza polypeptide (e.g., HA).
  • the linker may be a known linker sequence or may be generated de novo for use in the molecules described herein.
  • the VLP can be polyvalent, namely can include a plurality of influenza antigenic proteins, including any combination of wild-type and/or modified influenza polypeptides as described herein.
  • the transmembrane and/or cytoplasmic domain of the modified influenza polypeptide is replaced with a transmembrane and/or cytoplasmic domain from a different strain or subtype than the modified influenza polypeptide.
  • any of the VLPs described herein may further comprise additional influenza proteins, for example one or more additional wild-type matrix proteins (M1 and/or M2), one or more additional modified (mutant and/or hybrid) matrix proteins (M1 and/or M2), one or more wild-type antigenic glycoproteins (HA and/or NA), one or more hybrid antigenic glycoproteins (HA and/or NA), one or more modified antigenic glycoproteins (HA and/or NA), one or more hybrid and modified antigenic polypeptides (HA and/or NA), one or more nucleoproteins (NPs), one or more PB1 proteins, one or more PB2 proteins, one or more PA proteins and combinations thereof.
  • additional wild-type matrix proteins M1 and/or M2
  • additional modified (mutant and/or hybrid) matrix proteins M1 and/or M2
  • HA and/or NA wild-type antigenic glycoproteins
  • HA and/or NA hybrid antigenic glycoproteins
  • HA and/or NA one or more modified antigenic glycoproteins
  • NPs
  • the VLP is polyvalent in that more than one antigenic glycoprotein (any combination of modified or wild-type) is expressed on the surface of the VLP ( e . g ., to generate immune responses).
  • any of the VLPs described herein can be expressed in a eukaryotic cell (e . g ., a yeast cell, an insect cell, an amphibian cell, an avian cell, a plant cell or a mammalian cell) under conditions which permit the assembly and release of VLPs.
  • a host cell comprising any of the VLPs as described above.
  • the host cell permits assembly and release of a VLP as described herein from one or more vectors encoding the polypeptides of the VLP.
  • the eukaryotic cell is selected from the group consisting of a yeast cell, an insect cell, an amphibian cell, an avian cell, a plant cell or a mammalian cell.
  • a method of producing any of the VLPs described herein comprising the steps of transfecting one or more vectors encoding at least one matrix protein and at least one modified influenza hemagglutinin (HA) polypeptide comprising a hemagglutinin1 fragment and a hemagglutinin2 fragment operably linked to each other, into a suitable host cell, wherein the modification comprises a deletion of at least 150 amino acid residues of a hemagglutinin1 polypeptide, wherein the hemagglutinin2 fragment comprises the transmembrane and cytoplasmic tail domain of the hemagglutinin polypeptide and wherein the modification comprises mutations in one or more amino acids of a hemagglutinin2 polypeptide and expressing the combination of protein under conditions that allow VLP formation.
  • HA hemagglutinin
  • the matrix protein can be an influenza M1 protein, a thogoto matrix protein and an RSV matrix protein.
  • additional or the same vectors may encode additional proteins, for example additional influenza proteins (e.g., NA proteins, etc.).
  • the at least one vector further comprises a sequence encoding an influenza M2 protein.
  • the expression vector may be a plasmid, a viral vector, a baculovirus vector or a non-viral vector.
  • the vectors may encode one, more than one or all of the proteins of the VLP. In any of the methods of VLP production described herein, one or more of the vectors can be stably transfected into the host cell.
  • the at least one matrix protein and the modified influenza HA polypeptide are encoded on separate vectors and the vector encoding the at least one matrix protein is stably transfected into the cell prior to transfection with the vector encoding the modified influenza HA polypeptide protein.
  • the proteins encoded by the vectors may be full-length wild-type, full-length mutants, truncated wild-type, truncated mutants, and/or hybrid proteins include full-length and/or truncated wild-type or mutant proteins.
  • the cell can be a eukaryotic cell, for example, a yeast cell, an insect cell, an amphibian cell, an avian cell, a plant cell or a mammalian cell.
  • at least one M protein comprises an influenza matrix protein.
  • an immunogenic composition comprising at least one VLP as described herein.
  • the immunogenic composition further comprises an adjuvant.
  • Any of the immunogenic compositions described herein may include at least two VLPs comprising different modified influenza (e.g., HA) polypeptides.
  • the composition comprises and contains at least two VLPs, each VLP comprising a different modified HA protein or a single VLP displaying two or more remodeled HA or NA glycoproteins on its surface ( e . g .,. polyvalent VLP).
  • the immunogenic composition further comprises an adjuvant.
  • a method of generating an immune response to influenza in a subject comprising administering to the subject (e.g., human) an effective amount of a VLP and/or immunogenic composition as described herein to the subject.
  • the composition is administered mucosally, intradermally, subcutaneously, intramuscularly, or orally.
  • the methods generate an immune response to multiple strains or subtypes of influenza, thereby providing a "universal" vaccine that protects the subject against influenza infection from various influenza viruses and/or over time (more than one flu season).
  • Any of the methods may involve multiple administrations (e.g., a multiple dose schedule).
  • a packaging cell line for producing influenza VLPs as described herein.
  • the cell line is stably transfected with one or more polynucleotides encoding at least two M proteins and upon introduction and expression of the one or more influenza protein-encoding sequences not stably transfected into the cell, the VLP is produced by the cell.
  • sequences encoding M1 and/or M2 are stably integrated into the packaging cell line and sequences encoding the modified HA proteins expressed on the surface of the VLP are introduced into the cell such that the VLP is formed.
  • sequences encoding one or more of the modified HA proteins are stably integrated into the cell to form a packaging cell line and VLPs are formed upon introduction of sequences encoding the at least two M proteins.
  • the packaging cell may be an insect, plant, mammalian, bacterial or fungal cell. In certain embodiments, the packaging cell is a mammalian (e.g., human) cell line.
  • VLP virus-like particle
  • VLPs are generally composed of one or more viral proteins, such as, but not limited to those proteins referred to as capsid, coat, shell, surface and/or envelope proteins, or particle-forming polypeptides derived from these proteins. VLPs can form spontaneously upon recombinant expression of the protein in an appropriate expression system. Methods for producing particular VLPs are known in the art and discussed more fully below. The presence of VLPs following recombinant expression of viral proteins can be detected using conventional techniques known in the art, such as by electron microscopy, biophysical characterization, and the like.
  • VLPs can be isolated by density gradient centrifugation and/or identified by characteristic density banding (e.g., Examples) .
  • cryoelectron microscopy can be performed on vitrified aqueous samples of the VLP preparation in question, and images recorded under appropriate exposure conditions. Additional methods of VLP purification include but are not limited to chromatographic techniques such as affinity, ion exchange, size exclusion, and reverse phase procedures.
  • hybrid refers to a molecule (e.g., protein or VLP) that contains portions thereof, from at least two different proteins.
  • a hybrid influenza HA protein refers to a protein comprising at least a portion of an influenza HA protein (for example a portion containing one or more antigenic determinants) and portions of a heterologous protein (e.g., the cytoplasmic and/or transmembrane domain of a different influenza protein or a different viral protein, for example an RSV or VSV protein).
  • hybrid molecule as described herein can include full-length proteins fused to additional heterologous polypeptides (full length or portions thereof) as well as portions proteins fused to additional heterologous polypeptides (full length or portions thereof). It will also be apparent that the hybrids can include wild-type sequences or mutant sequences in any one, some or all of the heterologous domains.
  • polypeptide derived from a particular viral protein is meant a full-length or near full-length viral protein, as well as a fragment thereof, or a viral protein with internal deletions, which has the ability to form VLPs under conditions that favor VLP formation.
  • the polypeptide may comprise the full-length sequence, fragments, truncated and partial sequences, as well as analogs and precursor forms of the reference molecule. The term therefore intends deletions, additions and substitutions to the sequence, so long as the polypeptide retains the ability to form a VLP.
  • the term includes natural variations of the specified polypeptide since variations in coat proteins often occur between viral isolates.
  • substitutions are those which are conservative in nature, i.e., those substitutions that take place within a family of amino acids that are related in their side chains.
  • amino acids are generally divided into four families: (1) acidic--aspartate and glutamate; (2) basic--lysine, arginine, histidine; (3) nonpolar--alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar--glycine, asparagine, glutamine, cysteine, serine threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified as aromatic amino acids.
  • an “antigen” refers to a molecule containing one or more epitopes (either linear, conformational or both) that will stimulate a host's immune-system to make a humoral and/or cellular antigen-specific response.
  • the term is used interchangeably with the term "immunogen.”
  • a B-cell epitope will include at least about 5 amino acids but can be as small as 3-4 amino acids.
  • a T-cell epitope, such as a CTL epitope will include at least about 7-9 amino acids, and a helper T-cell epitope at least about 12-20 amino acids.
  • an epitope will include between about 7 and 15 amino acids, such as, 9, 10, 12 or 15 amino acids.
  • polypeptides which include modifications, such as deletions, additions and substitutions (generally conservative in nature) as compared to a native sequence, so long as the protein maintains the ability to elicit an immunological response, as defined herein. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the antigens.
  • an "immunological response" to an antigen or composition is the development in a subject of a humoral and/or a cellular immune response to an antigen present in the composition of interest.
  • a “humoral immune response” refers to an immune response mediated by antibody molecules
  • a “cellular immune response” is one mediated by T-lymphocytes and/or other white blood cells.
  • CTL cytolytic T-cells
  • CTLs have specificity for peptide antigens that are presented in association with proteins encoded by the major histocompatibility complex (MHC) and expressed on the surfaces of cells.
  • MHC major histocompatibility complex
  • helper T-cells help induce and promote the destruction of intracellular microbes, or the lysis of cells infected with such microbes.
  • Another aspect of cellular immunity involves an antigen-specific response by helper T-cells.
  • Helper T-cells act to help stimulate the function, and focus the activity of, nonspecific effector cells against cells displaying peptide antigens in association with MHC molecules on their surface.
  • a "cellular immune response” also refers to the production of cytokines, chemokines and other such molecules produced by activated T-cells and/or other white blood cells, including those derived from CD4+ and CD8+ T-cells.
  • an immunological response may include one or more of the following effects: the production of antibodies by B-cells; and/or the activation of suppressor T-cells and/or ⁇ T-cells directed specifically to an antigen or antigens present in the composition or vaccine of interest.
  • These responses may serve to neutralize infectivity, and/or mediate antibody-complement, or antibody dependent cell cytotoxicity (ADCC) to provide protection to an immunized host.
  • ADCC antibody dependent cell cytotoxicity
  • Such responses can be determined using standard immunoassays and neutralization assays, well known in the art.
  • an "immunogenic composition” is a composition that comprises an antigenic molecule where administration of the composition to a subject results in the development in the subject of a humoral and/or a cellular immune response to the antigenic molecule of interest.
  • substantially purified general refers to isolation of a substance (compound, polynucleotide, protein, polypeptide, polypeptide composition) such that the substance comprises the majority percent of the sample in which it resides.
  • a substantially purified component comprises 50%, preferably 80%-85%, more preferably 90-95% of the sample.
  • Techniques for purifying polynucleotides and polypeptides of interest are well-known in the art and include, for example, ion-exchange chromatography, affinity chromatography and sedimentation according to density.
  • a “coding sequence” or a sequence which "encodes” a selected polypeptide is a nucleic acid molecule which is transcribed (in the case of DNA) and translated (in the case of mRNA) into a polypeptide in vivo when placed under the control of appropriate regulatory sequences (or “control elements”).
  • the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus.
  • a coding sequence can include, but is not limited to, cDNA from viral, prokaryotic or eukaryotic mRNA, genomic DNA sequences from viral or prokaryotic DNA, and even synthetic DNA sequences.
  • a transcription termination sequence may be located 3' to the coding sequence.
  • control elements include, but are not limited to, transcription promoters, transcription enhancer elements, transcription termination signals, polyadenylation sequences (located 3' to the translation stop codon), sequences for optimization of initiation of translation (located 5' to the coding sequence), and translation termination sequences, and/ or sequence elements controlling an open chromatin structure see e.g., McCaughan et al. (1995) PNAS USA 92:5431-5435 ; Kochetov et al (1998) FEBS Letts. 440:351-355 .
  • a "nucleic acid" molecule can include, but is not limited to, prokaryotic sequences, eukaryotic mRNA, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic (e.g., mammalian) DNA, and even synthetic DNA sequences.
  • the term also captures sequences that include any of the known base analogs of DNA and RNA.
  • “Operably linked” refers to an arrangement of elements wherein the components so described are configured so as to perform their usual function.
  • a given promoter operably linked to a coding sequence is capable of effecting the expression of the coding sequence when active.
  • the promoter need not be contiguous with the coding sequence, so long as it functions to direct the expression thereof.
  • intervening untranslated yet transcribed sequences can be present between the promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked" to the coding sequence.
  • Recombinant as used herein to describe a nucleic acid molecule means a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which, by virtue of its origin or manipulation: (1) is not associated with all or a portion of the polynucleotide with which it is associated in nature; and/or (2) is linked to a polynucleotide other than that to which it is linked in nature.
  • the term "recombinant” as used with respect to a protein or polypeptide means a polypeptide produced by expression of a recombinant polynucleotide.
  • Recombinant host cells refer to cells which can be, or have been, used as recipients for recombinant vectors or other transfer DNA, and include the progeny of the original cell which has been transfected. It is understood that the progeny of a single parental cell may not necessarily be completely identical in morphology or in genomic or total DNA complement to the original parent, due to accidental or deliberate mutation.
  • Progeny of the parental cell which are sufficiently similar to the parent to be characterized by the relevant property, such as the presence of a nucleotide sequence encoding a desired peptide, are included in the progeny intended by this definition, and are covered by the above terms.
  • similarity means the exact amino acid to amino acid comparison of two or more polypeptides at the appropriate place, where amino acids are identical or possess similar chemical and/or physical properties such as charge or hydrophobicity. A so-termed “percent similarity” then can be determined between the compared polypeptide sequences.
  • Techniques for determining nucleic acid and amino acid sequence identity also are well known in the art and include determining the nucleotide sequence of the mRNA for that gene (usually via a cDNA intermediate) and determining the amino acid sequence encoded thereby, and comparing this to a second amino acid sequence.
  • identity refers to an exact nucleotide to nucleotide or amino acid to amino acid correspondence of two polynucleotides or polypeptide sequences, respectively.
  • Two or more polynucleotide sequences can be compared by determining their "percent identity.”
  • Two or more amino acid sequences likewise can be compared by determining their "percent identity.”
  • the percent identity of two sequences, whether nucleic acid or peptide sequences is generally described as the number of exact matches between two aligned sequences divided by the length of the shorter sequence and multiplied by 100.
  • An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981 ). This algorithm can be extended to use with peptide sequences using the scoring matrix developed by Dayhoff, Atlas of Protein Sequences and Structure, M. O. Dayhoff ed., 5 suppl.
  • a “vector” is capable of transferring gene sequences to target cells (e.g., bacterial plasmid vectors, viral vectors, non-viral vectors, particulate carriers, and liposomes).
  • target cells e.g., bacterial plasmid vectors, viral vectors, non-viral vectors, particulate carriers, and liposomes.
  • vector construct e.g., bacterial plasmid vectors, viral vectors, non-viral vectors, particulate carriers, and liposomes.
  • vector construct e.g., bacterial plasmid vectors, viral vectors, non-viral vectors, particulate carriers, and liposomes.
  • expression vector e.g., bacterial plasmid vectors, viral vectors, non-viral vectors, particulate carriers, and liposomes.
  • gene transfer vector mean any nucleic acid construct capable of directing the expression of one or more sequences of interest in a host cell.
  • the term includes
  • subject any member of the subphylum chordata, including, without limitation, humans and other primates, including non-human primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like.
  • the term does not denote a particular age. Thus, both adult and newborn individuals are intended to be covered.
  • the system described above is intended for use in any of the above vertebrate species, since the immune systems of all of these vertebrates operate similarly.
  • pharmaceutically acceptable or “pharmacologically acceptable” is meant a material which is not biologically or otherwise undesirable, i.e., the material may be administered to an individual in a formulation or composition without causing any unacceptable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • treatment refers to any of (i) the prevention of infection or reinfection, as in a traditional vaccine, (ii) the reduction or elimination of symptoms, and (iii) the substantial or complete elimination of the pathogen in question. Treatment may be effected prophylactically (prior to infection) or therapeutically (following infection).
  • adjuvant refers to a compound that, when used in combination with a specific immunogen (e.g. a VLP) in a formulation, will augment or otherwise alter or modify the resultant immune response. Modification of the immune response includes intensification or broadening the specificity of either or both antibody and cellular immune responses. Modification of the immune response can also mean decreasing or suppressing certain antigen-specific immune responses.
  • a specific immunogen e.g. a VLP
  • Modification of the immune response includes intensification or broadening the specificity of either or both antibody and cellular immune responses. Modification of the immune response can also mean decreasing or suppressing certain antigen-specific immune responses.
  • an "effective dose” generally refers to that amount of VLPs of the invention sufficient to induce immunity, to prevent and/or ameliorate an infection or to reduce at least one symptom of an infection and/or to enhance the efficacy of another dose of a VLP.
  • An effective dose may refer to the amount of VLPs sufficient to delay or minimize the onset of an infection.
  • An effective dose may also refer to the amount of VLPs that provides a therapeutic benefit in the treatment or management of an infection. Further, an effective dose is the amount with respect to VLPs of the invention alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of an infection.
  • An effective dose may also be the amount sufficient to enhance a subject's (e.g., a human's) own immune response against a subsequent exposure to an infectious agent.
  • Levels of immunity can be monitored, e.g., by measuring amounts of neutralizing secretory and/or serum antibodies, e.g., by plaque neutralization, complement fixation, enzyme-linked immunosorbent, or microneutralization assay.
  • an "effective dose" is one that prevents disease and/or reduces the severity of symptoms.
  • an effective amount refers to an amount of VLPs necessary or sufficient to realize a desired biologic effect.
  • An effective amount of the composition would be the amount that achieves a selected result, and such an amount could be determined as a matter of routine experimentation by a person skilled in the art.
  • an effective amount for preventing, treating and/or ameliorating an infection could be that amount necessary to cause activation of the immune system, resulting in the development of an antigen specific immune response upon exposure to VLPs of the invention.
  • the term is also synonymous with "sufficient amount.”
  • multivalent refers to VLPs which have multiple antigenic proteins against multiple types or strains of infectious agents.
  • immune stimulator refers to a compound that enhances an immune response via the body's own chemical messengers (cytokines). These molecules comprise various cytokines, lymphokines and chemokines with immunostimulatory, immunopotentiating, and pro-inflammatory activities, such as interferons, interleukins (e.g., IL-1, IL-2, IL-3, IL-4, IL-12, IL-13); growth factors (e.g., granulocyte-macrophage (GM)-colony stimulating factor (CSF)); and other immunostimulatory molecules, such as macrophage inflammatory factor, Flt3 ligand, B7.1; B7.2, etc.
  • the immune stimulator molecules can be administered in the same formulation as VLPs of the invention, or can be administered separately. Either the protein or an expression vector encoding the protein can be administered to produce an immunostimulatory effect.
  • protection immune response refers to an immune response mediated by antibodies against an infectious agent, which is exhibited by a vertebrate (e.g., a human), that prevents or ameliorates an infection or reduces at least one symptom thereof.
  • VLPs of the invention can stimulate the production of antibodies that, for example, neutralize infectious agents, blocks infectious agents from entering cells, blocks replication of said infectious agents, and/or protect host cells from infection and destruction.
  • the term can also refer to an immune response that is mediated by T-lymphocytes and/or other white blood cells against an infectious agent, exhibited by a vertebrate (e.g., a human), that prevents or ameliorates influenza infection or reduces at least one symptom thereof.
  • antigenic formulation or “antigenic composition” refers to a preparation which, when administered to a vertebrate, e.g. a mammal, will induce an immune response.
  • the term "vaccine” refers to a formulation which contains VLPs of the present invention, which is in a form that is capable of being administered to a vertebrate and which induces a protective immune response sufficient to induce immunity to prevent and/or ameliorate an infection and/or to reduce at least one symptom of an infection and/or to enhance the efficacy of another dose of VLPs.
  • the vaccine comprises a conventional saline or buffered aqueous solution medium in which the composition of the present invention is suspended or dissolved.
  • the composition of the present invention can be used conveniently to prevent, ameliorate, or otherwise treat an infection.
  • the vaccine Upon introduction into a host, the vaccine is able to provoke an immune response including, but not limited to, the production of antibodies and/or cytokines and/or the activation of cytotoxic T cells, antigen presenting cells, helper T cells, dendritic cells and/or other cellular responses.
  • influenza VLPs that can be used to protect and/or treat humans from influenza infection.
  • a VLP influenza vaccine that induces a strong immunological protective response against a variety of distinct flu viruses by deletion of amino acid residues and peptide sequences which create or compose a part of the immunodominant and/or highly variable regions of the virus (e.g., antigenic portions). This allows for otherwise immunologically cryptic epitopes becoming dominant and highly immunogenic.
  • the DNA sequence coding for this glycoprotein is rearranged by truncations, insertions, mutations or combinations thereof creating an open reading frame (ORF) for the synthesis of a remodeled HA molecule.
  • ORF open reading frame
  • the VLPs described herein elicit a distinct immune response as compared to the response stimulated by the viral infection or immunization with different vaccine compositions or formulations, including previously described VLPs.
  • the broadly neutralizing response elicited by this VPL vaccine will provide heterosubtypic protection against multiple influenza virus strains or subtypes.
  • VLP virus-like particles
  • the present disclosure relates to influenza VLPs from the plasma membrane of eukaryotic cells, which VLPs carry on their surfaces modified antigenic influenza proteins.
  • This VLP alone or in combination with one or more additional VLPs and/or adjuvants, stimulates an immune response that protects against influenza infection.
  • the VLPs described herein (also called sub-viral structure vaccine (SVSV)) is typically composed of viral proteins produced from naturally occurring and/or mutated nucleic acid sequences of genes coding for matrix protein M (also known as M1) and, optionally, M2 protein.
  • the matrix protein M is a universal component for the formation of all possible polyvalent sub-viral structure vaccine combinations.
  • the M1 and M2 proteins may be derived from any virus.
  • the M1 and/or M2 protein of the VLP is derived from an influenza matrix protein.
  • the M1 and/or M2 protein of the VLP is derived from RSV or thogoto-virus.
  • the M1 and/or M2 proteins may be modified (mutated), for example as disclosed herein or in U.S. Patent Publications 2008/0031895 and 2009/0022762 .
  • Influenza proteins derived from the same or different families of enveloped viruses can be selected for incorporation onto the surface of the vaccine.
  • the incorporation of influenza proteins into the same vaccine particle can be facilitated by replacing the cytoplasmic tail and transmembrane amino acid sequences with those from a common glycoprotein via alterations in the nucleic acids coding for these proteins. This approach allows for the design of a large number of possible polyvalent sub-viral vaccine combinations.
  • VLPs described herein comprise modified (e.g., truncated, deletions, hybrid, etc.) influenza antigenic polypeptides.
  • the modified influenza antigen is a hemagglutinin (HA) polypeptide.
  • the major functions of the hemagglutinin (HA) molecule are receptor-binding and membrane fusion activity, critical steps in the initiation of viral infection.
  • HA is the major surface antigen of the virus against which neutralizing antibodies are produced.
  • the precursor molecule (HA0) is cleaved into two polypeptides (HA1) and (HA2) and covalently linked by a single disulphide bond. Cleavage of the HA0 activates the fusion peptide on the NH2 terminal of the HA2 subunit and activates the fusion potential of the HA, an essential process required for virus infectivity.
  • the most important and immunological dominant antigenic sites on the HA structure reside on the head of the molecule, primarily formed by the HA1 subunit. Changes in these antigenic regions by mutations (antigenic drift) or HA swapping (antigenic shift) allows for the virus to escape host neutralizing antibodies and initiate infection of new cells. In addition to these sites, subdominant and highly conserved epitopes are present on the stem portion of the structure; however, because of immunological hierarchy these sites are mainly unrecognized by the immune system and unable to generate a significant antibody response. Nonetheless, antibodies targeting these regions are able to block infection by inhibiting membrane fusion and therefore virus entry.
  • the vaccine described in this invention is based on expressing, in a VLP, truncated, re-engineered and remodeled HA molecules that lack the major immunodominant epitopes and predominantly display subdominant antigenic sites present on the stem portion of the molecule.
  • These structurally modified HAs are incorporated on the surface of virus-like particles (VLP) forming single noninfectious protective particle (SNIPP) influenza vaccines.
  • VLP virus-like particles
  • SNIPP single noninfectious protective particle
  • the modified (remodeled) HA polypeptides lack a portion of the HA1 domain, for example by deleting some or all of the immunodominant epitopes, while maintaining a small NH2 terminal region containing the translocation signal sequence which is linked to alternative configurations of HA1 and HA2 subunits.
  • the deletions (truncations) can be of any length, for example from 10 to 300 base pairs (or any value therebetween, for example, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300 or more base pairs).
  • deletions can be of consecutive base pairs or, alternatively, base pairs can be deleted in different regions of the HA polypeptide, for example deleting 10 or more base pairs at the N-terminal and additional base pairs (10 to 300 or even more) in the central region of the molecule ( e . g ., a region of 10 or more base pairs C-terminal to the translocation signal sequence). Additional modifications include, but are not limited to, addition of linkers, mutation of one or more residues of the polypeptide and the like. The specific modifications of the HA molecule can be readily performed at the DNA level and subsequently sub-clone together with the combination of genes required for VLP assembly.
  • antigenic influenza proteins e.g., HA
  • VLP can be modified in any way, for example via deletions of one or more amino acids and/or mutations of one amino acid.
  • An influenza HA polypeptide may be modified by deleting amino acids near the N-terminal of the HA polypeptide. For example, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33, the residues from H24 to G240 may be deleted. Optionally, other portions of the HA1 NH2 terminal are deleted while retaining amino acids D17 to Y23 and the secretion signal sequence. The retained small sequence maintains residue C20 which promotes the formation of an inter-chain disulfide bond with C483 and stabilizes the molecule. The remaining region of HA1 (R241-L333) forms a series of beta sheets interspersed with bend conserving residue C294 which forms a disulfide bond with C318. Optionally, a residue at position C290 is mutated to G to prevent potential disulfide bond formation with either C294 or C318.
  • a modified (remodeled) HA may comprise the NH2 terminal of HA1 (e.g., D17 to P65, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33) which includes residues that promote disulfide bonding (e.g. C20 and C483, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33).
  • Such modified HA polypeptides can be linked to additional regions of HA1, for examples residues 241 to 346, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33, which includes the enzymatic cleavage site and unchanged C290 to promote the formation of a disulfide bond with C58.
  • a modified HA that includes the NH2 terminal portion of the HA1 (D17 to P65, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33) is provided.
  • this fragment can be linked to another portion of the HA1 (e.g., M281-R346, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33), for example through a peptide linker. Any peptide linker of any length can be used.
  • the peptide linker is between 5 and 25 amino acids (including any number therebetween), for example a 12 amino acid linker (DIGPGKVGYGPG, SEQ ID NO:11).
  • the residues that promote the formation of disulfide bonds are maintained (e.g., C20-C483, C294-C318 and C58-C290 pairing residues, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33).
  • any of the modified HA1 polypeptides (fragments) described herein can be operably linked to an HA2 fragment (e.g., an HA2 fragment comprising residues G347-I568, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33)) comprising the transmembrane domain and cytoplasmic tail.
  • mutations may be introduced into one or more amino acids of the HA2 polypeptide, for example, mutations of residues that inhibit or prevent protein aggregation (e.g., V412 to D412 and L419 to G419, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33). See , e . g ., Figure 3 .
  • a modified HA may comprise of an entire HA2 fragment linked to the secretion signal peptide.
  • This construct comprises a secretion signal sequence, the HA2 extracellular (e . g ., 347-520, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33) transmembrane and cytoplasmic ( e . g ., 521-568, as numbered relative to wild-type HA sequence UniProt KB Q6DQ33) domains.
  • the signal peptide is removed during transcription/translocation and it is not present in the HA2 structure incorporated onto the VLP vaccine; as it is the case for the other remodeled HAs.
  • transmembrane domain and cytoplasmic tail at the carboxyl terminal of the HA2 subunits can be replaced with analogous sequences of an influenza virus from which the M1 protein is derived or the sequence that best interacts with an M1 analogue. See , e . g ., Figure 4 .
  • the HA may be modified by inserting two or more 12-mers multi basic proteolytic cleavage sites within the sequence of the protein.
  • suitable insertion sites include between P65 and L66 and/or between 1280 and M281.
  • one or more residues of the native proteolytic cleavage site (KKR to GGG) can also be mutated in order to prevent cleavage at this location. Proteases treatment of this molecule removes the globular head of the HA exposing the conserved epitopes formed by the HA2 stem and remaining portions of HA1. See , e.g., Figure 5A and B .
  • VLPs produced as described herein are conveniently prepared using standard recombinant techniques.
  • Polynucleotides encoding the VLP-forming protein(s) are introduced into a host cell and, when the proteins are expressed in the cell, they assembly into VLPs.
  • Polynucleotide sequences coding for molecules (structural and/or antigen polypeptides, including modified antigenic (e.g., HA) polypeptides) that form and/or are incorporated into the VLPs can be obtained using recombinant methods, such as by screening cDNA and genomic libraries from cells expressing the gene, or by deriving the gene from a vector known to include the same.
  • plasmids which contain sequences that encode naturally occurring or altered cellular products may be obtained from a depository such as the A.T.C.C., or from commercial sources.
  • Plasmids containing the nucleotide sequences of interest can be digested with appropriate restriction enzymes, and DNA fragments containing the nucleotide sequences can be inserted into a gene transfer vector using standard molecular biology techniques.
  • cDNA sequences may be obtained from cells which express or contain the sequences, using standard techniques, such as phenol extraction and PCR of cDNA or genomic DNA. See, e.g., Sambrook et al., supra, for a description of techniques used to obtain and isolate DNA. Briefly, mRNA from a cell which expresses the gene of interest can be reverse transcribed with reverse transcriptase using oligo-dT or random primers. The single stranded cDNA may then be amplified by PCR (see U.S. Pat. Nos.
  • the nucleotide sequence of interest can also be produced synthetically, rather than cloned, using a DNA synthesizer (e.g., an Applied Biosystems Model 392 DNA Synthesizer, available from ABI, Foster City, Calif.).
  • the nucleotide sequence can be designed with the appropriate codons for the expression product desired.
  • the complete sequence is assembled from overlapping oligonucleotides prepared by standard methods and assembled into a complete coding sequence. See, e.g., Edge (1981) Nature 292:756 ; Nambair et al. (1984) Science 223:1299 ; Jay et al. (1984) J. Biol. Chem. 259:6311 .
  • the VLPs described herein are typically formed by expressing sequences encoding M1 and at least one modified influenza (e.g., HA antigen) in a cell, optionally with an M2 protein.
  • the expressed proteins self-assemble into VLPs with the antigenic glycoproteins decorating the surface of the VLP.
  • the matrix-encoding sequences may be RSV matrix proteins.
  • the matrix-encoding sequences may be influenza matrix proteins. It will also be apparent that the matrix-encoding sequences can contain one or more mutations (modifications), for example the modified matrix proteins as described in U.S. Patent Publications 2008/0031895 and 2009/0022762 .
  • the VLPs described herein may further comprise additional influenza proteins (wild-type, modified (mutants) and/or hybrids of wild-type or mutants).
  • any of the proteins used in the VLPs described herein may be hybrid (or chimeric) proteins. It will be apparent that all or parts of the polypeptides may be replaced with sequences from other viruses and/or sequences from other influenza strains. According to the disclosure, any of the proteins of the VLP may be hybrids in that they include heterologous sequences encoding the transmembrane and/or cytoplasmic tail domains, for example domains from influenza proteins such as HA or NA. See, e.g., U.S. Patent Publication Nos. 2008/0031895 and 2009/0022762 .
  • the sequences employed to form influenza VLPs exhibit between about 60% to 80% (or any value therebetween including 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78% and 79%) sequence identity to a naturally occurring influenza polynucleotide sequence and more preferably the sequences exhibit between about 80% and 100% (or any value therebetween including 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99%) sequence identity to a naturally occurring polynucleotide sequence.
  • any of the sequences described herein may further include additional sequences.
  • hybrid molecules are expressed and incorporated into the sub-viral structure. These hybrid molecules are generated by linking, at the DNA level, the sequences coding for the matrix protein genes with sequences coding for an adjuvant or immuno-regulatory moiety. During sub-viral structure formation, these hybrid proteins are incorporated into or onto the particle depending on whether M1 or optional M2 carries the adjuvant molecule.
  • the incorporation of one or more polypeptide immunomodulatory polypeptides e.g., adjuvants describe in detail below
  • one or more additional molecules may be included in the VLP-containing compositions after production of the VLP from the sequences described herein.
  • sequences described herein can be operably linked to each other in any combination.
  • one or more sequences may be expressed from the same promoter and/or from different promoters.
  • sequences may be included on one or more vectors.
  • constructs comprising the sequences encoding the polypeptide(s) desired to be incorporated into the VLP
  • they can be cloned into any suitable vector or replicon for expression.
  • Numerous cloning vectors are known to those of skill in the art, and one having ordinary skill in the art can readily select appropriate vectors and control elements for any given host cell type in view of the teachings of the present specification and information known in the art about expression. See, generally, Ausubel et al, supra or Sambrook et al, supra.
  • Non-limiting examples of vectors that can be used to express sequences that assembly into VLPs as described herein include viral-based vectors (e.g., retrovirus, adenovirus, adeno-associated virus, lentivirus), baculovirus vectors (see, Examples), plasmid vectors, non-viral vectors, mammalians vectors, mammalian artificial chromosomes (e.g., liposomes, particulate carriers, etc.) and combinations thereof.
  • viral-based vectors e.g., retrovirus, adenovirus, adeno-associated virus, lentivirus
  • baculovirus vectors see, Examples
  • plasmid vectors e.g., retrovirus, adenovirus, adeno-associated virus, lentivirus
  • baculovirus vectors see, Examples
  • plasmid vectors e.g., non-viral vectors
  • mammalians vectors e.g., mam
  • the expression vector(s) typically contain(s) coding sequences and expression control elements which allow expression of the coding regions in a suitable host.
  • the control elements generally include a promoter, translation initiation codon, and translation and transcription termination sequences, and an insertion site for introducing the insert into the vector.
  • Translational control elements have been reviewed by M. Kozak (e.g., Kozak, M., Mamm. Genome 7(8):563-574, 1996 ; Kozak, M., Biochimie 76(9):815-821, 1994 ; Kozak, M., J Cell Biol 108(2):229-241, 1989 ; Kozak, M., and Shatkin, A. J., Methods Enzymol 60:360-375, 1979 ).
  • typical promoters for mammalian cell expression include the SV40 early promoter, a CMV promoter such as the CMV immediate early promoter (a CMV promoter can include intron A), RSV, HIV-LTR, the mouse mammary tumor virus LTR promoter (MMLV-LTR), FIV-LTR, the adenovirus major late promoter (Ad MLP), and the herpes simplex virus promoter, among others.
  • CMV promoter such as the CMV immediate early promoter
  • RSV CMV immediate early promoter
  • HIV-LTR the mouse mammary tumor virus LTR promoter
  • MMLV-LTR mouse mammary tumor virus LTR promoter
  • FIV-LTR the adenovirus major late promoter
  • Ad MLP adenovirus major late promoter
  • Other nonviral promoters such as a promoter derived from the murine metallothionein gene, will also find use for mammalian expression.
  • transcription termination and polyadenylation sequences will also
  • a sequence for optimization of initiation of translation located 5' to the coding sequence, is also present.
  • transcription terminator/polyadenylation signals include those derived from SV40, as described in Sambrook, et al., supra, as well as a bovine growth hormone terminator sequence.
  • Introns, containing splice donor and acceptor sites, may also be designed into the constructs as described herein ( Chapman et al., Nuc. Acids Res. (1991) 19:3979-3986 ).
  • Enhancer elements may also be used herein to increase expression levels of the mammalian constructs. Examples include the SV40 early gene enhancer, as described in Dijkema et al., EMBO J. (1985) 4:761 , the enhancer/promoter derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus, as described in Gorman et al., Proc. Natl. Acad. Sci. USA (1982b) 79:6777 and elements derived from human CMV, as described in Boshart et al., Cell (1985) 41:521 , such as elements included in the CMV intron A sequence ( Chapman et al., Nuc. Acids Res. (1991) 19:3979-3986 ).
  • LTR long terminal repeat
  • one or more vectors may contain one or more sequences encoding proteins to be incorporated into the VLP.
  • a single vector may carry sequences encoding all the proteins found in the VLP.
  • multiple vectors may be used ( e . g ., multiple constructs, each encoding a single polypeptide-encoding sequence or multiple constructs, each encoding one or more polypeptide-encoding sequences).
  • the sequences may be operably linked to the same or different transcriptional control elements (e . g ., promoters) within the same vector.
  • vectors may contain additional gene expression controlling sequences including chromatin opening elements which prevent transgene silencing and confer consistent, stable and high level of gene expression, irrespective of the chromosomal integration site.
  • chromatin opening elements located in proximity of house-keeping genes, which in the vectors create a transcriptionally active open chromatin environment around the integrated transgene, maximizing transcription and protein expression, irrespective of the position of the transgene in the chromosome.
  • sequences encoding non-influenza proteins may be expressed and incorporated into the VLP, including, but not limited to, sequences comprising and/or encoding immunomodulatory molecules (e.g., adjuvants described below), for example, immunomodulating oligonucleotides (e.g., CpGs), cytokines, detoxified bacterial toxins and the like.
  • immunomodulatory molecules e.g., adjuvants described below
  • immunomodulating oligonucleotides e.g., CpGs
  • cytokines detoxified bacterial toxins and the like.
  • influenza proteins expressed in a eukaryotic host cell have been shown to self-assemble into noninfectious virus-like particles (VLP). Accordingly, the sequences and/or vectors described herein are then used to transform an appropriate host cell.
  • the construct(s) encoding the proteins that form the VLPs described herein provide efficient means for the production of influenza VLPs using a variety of different cell types, including, but not limited to, insect, fungal (yeast) and mammalian cells.
  • the sub-viral structure vaccines are produced in eukaryotic cells following transfection, establishment of continuous cell lines (using standard protocols) and/or infection with DNA constructs that carry the influenza genes of interest as known to one skilled in the art.
  • the level of expression of the proteins required for sub-viral structure formation is maximized by sequence optimization of the eukaryotic or viral promoters that drive transcription of the selected genes.
  • the sub-viral structure vaccine is released into the culture media, from where it is purified and subsequently formulated as a vaccine.
  • the sub-viral structures are not infectious and therefore inactivation of the VLP is not required as it is for some killed viral vaccines
  • influenza polypeptides expressed from sequences as described herein to self-assemble into VLPs with antigenic glycoproteins presented on the surface allows these VLPs to be produced in many host cell by co-introduction of the desired sequences.
  • the sequence(s) e.g., in one or more expression vectors
  • Suitable host cells include, but are not limited to, bacterial, mammalian, baculovirus/insect, yeast, plant and Xenopus cells.
  • mammalian cell lines include primary cells as well as immortalized cell lines available from the American Type Culture Collection (A.T.C.C.), such as, but not limited to, MDCK, BHK, VERO, MRC-5, WI-38, HT1080, 293, 293T, RD, COS-7, CHO, Jurkat, HUT, SUPT, C8166, MOLT4/clone8, MT-2, MT-4, H9, PM1, CEM, myeloma cells (e.g., SB20 cells) and CEMX174 (such cell lines are available, for example, from the A.T.C.C.).
  • A.T.C.C. American Type Culture Collection
  • bacterial hosts such as E . coli , Bacillus subtilis , and Streptococcus spp ., will find use with the present expression constructs.
  • Yeast hosts useful in the present disclosure include inter alia , Saccharomyces cerevisiae, Candida albicans, Candida maltosa, Hansenula polymorpha, Kluyveromyces fragilis, Kluyveromyces lactis, Pichia guillerimondii , Pichia pastoris , Schizosaccharomycespombe and Yarrowia lipolytica .
  • Fungal hosts include, for example, Aspergillus .
  • Insect cells for use with baculovirus expression vectors include, inter alia, Aedes aegypti, Autographa californica , Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda , and Trichoplusia ni . See, Latham & Galarza (2001) J. Virol. 75(13):6154-6165 ; Galarza et al. (2005) Viral. Immunol. 18(1):244-51 ; and U.S. Patent Publications 200550186621 and 20060263804 .
  • Cell lines expressing one or more of the sequences described above can readily be generated given the disclosure provided herein by stably integrating one or more expression vector constructs encoding the proteins of the VLP.
  • the promoter regulating expression of the stably integrated influenza sequences (s) may be constitutive or inducible.
  • a cell line can be generated in which one or more both of the matrix proteins are stably integrated such that, upon introduction of the sequences described herein (e . g ., hybrid proteins) into a host cell and expression of the proteins encoded by the polynucleotides, non-replicating viral particles that present antigenic glycoproteins are formed.
  • a mammalian cell line that stably expressed two or more antigenically distinct influenza proteins is generated. Sequences encoding M1, M2 and/or additional glycoproteins (e.g., from the same or different virus strains) can be introduced into such a cell line to produce VLPs as described herein. Alternatively, a cell line that stably produces an M1 protein (and, optionally, M2) can be generated and sequences encoding the antigenic influenza protein(s) from the selected strain(s) introduced into the cell line, resulting in production of VLPs presenting the desired antigenic glycoproteins.
  • the parent cell line from which an VLP-producer cell line is derived can be selected from any cell described above, including for example, mammalian, insect, yeast, bacterial cell lines.
  • the cell line is a mammalian cell line (e.g., 293, RD, COS-7, CHO, BHK, MDCK, MDBK, MRC-5, VERO, HT1080, and myeloma cells).
  • Production of influenza VLPs using mammalian cells provides (i) VLP formation; (ii) correct post translation modifications (glycosylation, palmitylation) and budding; (iii) absence of non-mammalian cell contaminants and (iv) ease of purification.
  • influenza-encoding sequences may also be transiently expressed in host cells.
  • Suitable recombinant expression host cell systems include, but are not limited to, bacterial, mammalian, baculovirus/insect, vaccinia, Semliki Forest virus (SFV), Alphaviruses (such as, Sindbis, Venezuelan Equine Encephalitis (VEE)), mammalian, yeast and Xenopus expression systems, well known in the art.
  • Particularly preferred expression systems are mammalian cell lines, vaccinia, Sindbis, insect and yeast systems.
  • baculovirus expression Reilly, P. R., et al., BACULOVIRUS EXPRESSION VECTORS: A LABORATORY MANUAL (1992 ); Beames, et al., Biotechniques 11:378 (1991 ); Pharmingen; Clontech, Palo Alto, Calif.)
  • vaccinia expression systems Earl, P. L., et al., "Expression of proteins in mammalian cells using vaccinia" In Current Protocols in Molecular Biology (F. M. Ausubel, et al.
  • Plant cloning vectors Clontech Laboratories, Inc., Palo-Alto, Calif., and Pharmacia LKB Biotechnology, Inc., Pistcataway, N.J.; Hood, E., et al., J. Bacteriol. 168:1291-1301 (1986 ); Nagel, R., et al., FEMS Microbiol. Lett. 67:325 (1990 ); An, et al., "Binary Vectors", and others in Plant Molecular Biology Manual A3:1-19 (1988 ); Miki, B. L.
  • the VLPs are produced by growing host cells transformed by an expression vector under conditions whereby the particle-forming polypeptide(s) is(are) expressed and VLPs can be formed.
  • the selection of the appropriate growth conditions is within the skill of the art. If the VLPs are formed and retained intracellularly, the cells are then disrupted, using chemical, physical or mechanical means, which lyse the cells yet keep the VLPs substantially intact. Such methods are known to those of skill in the art and are described in, e.g., Protein Purification Applications: A Practical Approach, (E. L. V. Harris and S. Angal, Eds., 1990 ). Alternatively, VLPs may be secreted and harvested from the surrounding culture media.
  • the particles are then isolated (or substantially purified) using methods that preserve the integrity thereof, such as, by density gradient centrifugation, e.g., sucrose gradients, PEG-precipitation, pelleting, and the like (see, e.g., Kirnbauer et al. J. Virol. (1993) 67:6929-6936 ), as well as standard purification techniques including, e.g., ion exchange and gel filtration chromatography.
  • density gradient centrifugation e.g., sucrose gradients, PEG-precipitation, pelleting, and the like
  • standard purification techniques including, e.g., ion exchange and gel filtration chromatography.
  • VLPs produced as described herein can be used to elicit an immune response when administered to a subject.
  • the VLPs can comprise a variety of antigens (e.g., one or more modified influenza antigens from one or more strains or isolates).
  • Purified VLPs can be administered to a vertebrate subject, usually in the form of vaccine compositions.
  • Combination vaccines may also be used, where such vaccines contain, for example, other subunit proteins derived from influenza or other organisms and/or gene delivery vaccines encoding such antigens.
  • VLP immune-stimulating (or vaccine) compositions can include various excipients, adjuvants, carriers, auxiliary substances, modulating agents, and the like.
  • the immune stimulating compositions will include an amount of the VLP/antigen sufficient to mount an immunological response.
  • An appropriate effective amount can be determined by one of skill in the art. Such an amount will fall in a relatively broad range that can be determined through routine trials and will generally be an amount on the order of about 0.1 ⁇ g to about 10 (or more) mg, more preferably about 1 ⁇ g to about 300 ⁇ g, of VLP/antigen.
  • Sub-viral structure vaccines are purified from the cell culture media and formulated with the appropriate buffers and additives, such as a) preservatives or antibiotics; b) stabilizers, including proteins or organic compounds; c) adjuvants or immuno-modulators for enhancing potency and modulating immune responses (humoral and cellular) to the vaccine; or d) molecules that enhance presentation of vaccine antigens to specifics cell of the immune system.
  • This vaccine can be prepared in a freeze-dried (lyophilized) form in order to provide for appropriate storage and maximize the shelf-life of the preparation. This will allow for stock piling of vaccine for prolonged periods of time maintaining immunogenicity, potency and efficacy.
  • a carrier is optionally present in the compositions described herein.
  • a carrier is a molecule that does not itself induce the production of antibodies harmful to the individual receiving the composition.
  • Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycollic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles.
  • particulate carriers include those derived from polymethyl methacrylate polymers, as well as microparticles derived from poly(lactides) and poly(lactide-co-glycolides), known as PLG. See, e.g., Jeffery et al., Pharm. Res.
  • these carriers may function as immunostimulating agents ("adjuvants").
  • adjuvants include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc.; (2) oil-in-water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components), such as for example (a) MF59 (International Publication No.
  • WO 90/14837 containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE (see below), although not required) formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton, Mass.), (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP (see below) either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS),
  • cytokines such as interleukins (IL-1, IL-2, etc.), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), beta chemokines (MIP, 1-alpha, 1-beta Rantes, etc.
  • cytokines such as interleukins (IL-1, IL-2, etc.), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), beta chemokines (MIP, 1-alpha, 1-beta Rantes, etc.
  • cytokines such as interleukins (IL-1, IL-2, etc.
  • M-CSF macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • MIP beta chemokines
  • coli heat-labile toxin particularly LT-K63 (where lysine is substituted for the wild-type amino acid at position 63)
  • LT-R72 where arginine is substituted for the wild-type amino acid at position 72
  • CT-S109 where serine is substituted for the wild-type amino acid at position 109
  • PT-K9/G129 where lysine is substituted for the wild-type amino acid at position 9 and glycine substituted at position 129)
  • LT heat-labile toxin
  • CT-S109 where serine is substituted for the wild-type amino acid at position 109
  • PT-K9/G129 where lysine is substituted for the wild-type amino acid at position 9 and glycine substituted at position 129
  • Muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acteyl-normuramyl-L-alanyl-D-isogluatme (nor-MDP), N-acetylmuramyl-L-alanyl-D-isogluatminyl-L-alanine-2-(1'-2'-dipalmitoyl-sn -glycero-3-huydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
  • thr-MDP N-acetyl-muramyl-L-threonyl-D-isoglutamine
  • nor-MDP N-acteyl-normuramyl-L-alanyl-D-isogluatme
  • MTP-PE N-acetylmuramyl-L-
  • immunomodulatory molecules for use herein include adjuvants described above and the following: IL-1 and IL-2 ( Karupiah et al. (1990) J. Immunology 144:290-298 , Weber et al. (1987) J. Exp. Med. 166:1716-1733 , Gansbacher et al. (1990) J. Exp. Med. 172:1217-1224 , and U.S. Patent No. 4,738,927- ); IL-3 and IL-4 ( Tepper et al. (1989) Cell 57:503-512 , Golumbek et al. (1991) Science 254:713-716 , and U.S. Patent No.
  • IL-5 and IL-6 Brakenhof et al. (1987) J. Immunol. 139:4116-4121 , and International Publication No. WO 90/06370
  • IL-7 U.S. Pat. No. 4,965,195
  • IL-8, IL-9, IL-10, IL-11, IL-12, and IL-13 Cytokine Bulletin, Summer 1994
  • IL-14 and IL-15 alpha interferon ( Finter et al. (1991) Drugs 42:749-765 , U.S. Pat. Nos. 4,892,743 and 4,966,843 , International Publication No. WO 85/02862 , Nagata et al.
  • Immunomodulatory factors may also be agonists, antagonists, or ligands for these molecules. For example, soluble forms of receptors can often behave as antagonists for these types of factors, as can mutated forms of the factors themselves.
  • Nucleic acid molecules that encode the above-described substances, as well as other nucleic acid molecules that are advantageous for use within the present invention may be readily obtained from a variety of sources, including, for example, depositories such as the American Type Culture Collection, or from commercial sources such as British Bio-Technology Limited (Cowley, Oxford England). Representative examples include BBG 12 (containing the GM-CSF gene coding for the mature protein of 127 amino acids), BBG 6 (which contains sequences encoding gamma interferon), A.T.C.C. Deposit No. 39656 (which contains sequences encoding TNF), A.T.C.C. Deposit No. 20663 (which contains sequences encoding alpha-interferon), A.T.C.C.
  • Plasmids encoding one or more of the above-identified polypeptides can be digested with appropriate restriction enzymes, and DNA fragments containing the particular gene of interest can be inserted into a gene transfer vector (e . g ., expression vector as described above) using standard molecular biology techniques. (See, e.g., Sambrook et al., supra , or Ausubel et al. (eds) Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience ).
  • VLPs and compositions comprising these VLPs can be administered to a subject by any mode of delivery, including, for example, by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (e.g. see WO99/27961 ) or transcutaneous (e.g. see WO02/074244 and WO02/064162 ), intranasal (e.g. see WO03/028760 ), ocular, aural, pulmonary or other mucosal administration. Multiple doses can be administered by the same or different routes. The doses may be intranasally administered.
  • parenteral injection e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue
  • rectal e.g. tablet, spray
  • VLPs and VLP-containing compositions
  • the site of VLP administration may be the same or different as other vaccine compositions that are being administered.
  • Dosage treatment with the VLP composition may be a single dose schedule or a multiple dose schedule.
  • a multiple dose schedule is one in which a primary course of vaccination may be with 1-10 separate doses, followed by other doses given at subsequent time intervals, chosen to maintain and/or reinforce the immune response, for example at 1-4 months for a second dose, and if needed, a subsequent dose(s) after several months.
  • the dosage regimen will also, at least in part, be determined by the potency of the modality, the vaccine delivery employed, the need of the subject and be dependent on the judgment of the practitioner.
  • Example 1 Generation of truncated, re-engineered and remodeled HA molecules
  • a remodeled HA molecule was generated by deleting a portion (217aa, from H23 to G240) of the HA1 fragment, maintaining amino acids D17 to Y23 at the NH2 terminal after removal of the secretion signal sequence.
  • This small sequence maintained residue C20 which promotes the formation of an inter-chain disulfide bond with C483 and stabilizes the molecule.
  • the remaining region of HA1 R241-L333 forms a series of beta sheets interspersed with bend conserving residue C294 which forms a disulfide bond with C318.
  • a residue at position C290 was mutated to G to prevent potential disulfide bond formation with either C294 or C318.
  • This HA1 fragment was genetically linked to the entire HA2 fragment comprising the transmembrane domain and cytoplasmic tail.
  • HA1 Another remodeled HA was designed to encompass the NH2 terminal of HA1 (D17 to P65) which includes C20 to promote disulfide bond with C483 and a portion that contributes to the formation of highly conserved epitope. This was linked to remaining portion of HA1, position 241 to 346, which includes the enzymatic cleavage site and unchanged C290 to promote the formation of a disulfide bond with C58.
  • This HA1 fragment was genetically linked to the entire HA2 fragment (G347-1568) comprising the transmembrane and cytoplasmic domains.
  • This construct was designated as remodeled HA "2TA” and depicted in Figure 2 .
  • the DNA and amino acid sequences are shown below.
  • FIG. 3 Another remodeled HA is shown in Figure 3 .
  • This construct includes the NH2 terminal portion of the HA1 (D17 to P65), after the cleavage of the signal peptide, linked to another portion of the HA1 (M281-R346) through a designed 12aa peptide linker (DIGPGKVGYGPG, SEQ ID NO:11).
  • DIGPGKVGYGPG 12aa peptide linker
  • This construct is identified as "3TA remodeled HA" and a schematic is shown in Figure 3 .
  • the nucleotide and amino acid sequence of the 3TA molecule is shown below.
  • FIG. 4 Another example of a modified influenza HA is shown in Figure 4 (designated "4TA").
  • This construct includes an HA2 fragment (including the extracellular region (residues 347-520) and transmembrane and cytoplasmic domains (residues 521-568) domains linked to the secretion signal peptide.
  • the signal peptide is removed during transcription/translocation and it is not present in the HA2 structure incorporated onto the VLP vaccine (as is the case for the other remodeled HAs).
  • transmembrane domain and cytoplasmic tail at the carboxyl terminal of the HA2 subunits are optionally replaced with analogous sequences of an influenza virus from which the M1 protein is derived or the sequence that best interacts with an M1 analogue. Optimizing these molecular contacts enhances particle assembly and release.
  • the nucleotide and amino acid sequence of the 4TA molecule is shown below.
  • an influenza HA polypeptide is remodeled by inserting one or more cleavage sites within the sequence of the protein.
  • exemplary insertion sites include is between P65 and L66 and/or between 1280 and M281.
  • Figure 5 also shows an exemplary cleavage site of 12 residues (e.g., 12-mers multi basic proteolytic cleavage sites).
  • An additional change includes mutation of 3 residues in the native proteolytic cleavage site (KKR to GGG) can also be included in order to prevent cleavage at this location.
  • Proteases treatment of this molecule removes the globular head of the HA exposing the conserved epitopes formed by the HA2 stem and remaining portions of HA1.
  • the nucleotide and amino acid sequences of the 5TA molecule are shown below.
  • remodeled structures typically lack a large portion of the HA1 domain, deleting the majority of the immunodominant epitopes but maintaining a small NH2 terminal region containing the translocation signal sequence which is linked to alternative configurations of HA1 and HA2 subunits.
  • the specific modifications of the HA molecule are performed at the DNA level and subsequently sub-clone together with the combination of genes required for VLP assembly.
  • VLPs virus-like particles
  • VLPs As described in detail above, a variety of ways to produce and evaluate the appropriate VLPs are known to one knowledgeable in the art.
  • the genes required for VLP formation including the DNA sequences coding for the modified HA can be expressed in eukaryotic cells.
  • Alternative recombinant expression systems are used for the production of VLPs which include but are not limited to baculovirus/insect cell systems, transiently or stably transfected mammalian, yeast, plant cells or prokaryotic cells using plasmids, vectors, viruses or other methods for the introduction and expression of single or multiple genes into cells.
  • VLPs are tested for reactivity with antibodies that recognize immunodominant and subdominant regions.
  • VLP structures carrying modified HAs that react with antibodies recognizing conserved subdominant epitopes are used to immunize animals and evaluate the capacity of the sera to neutralize wild type intact virus as well as the ability to elicit a broadly protective immune response against challenge viruses in mice and ferrets.
  • VLP vaccine formulated with VLPs assembled with modified HAs as described herein of type A and B viruses
  • VLP vaccine will elicit a broadly neutralizing immune response capable of protecting against subtypes and antigenic variants of the type A as well as type B influenza viruses and their antigenic variants.
  • Example 2 Generation Of Mammalian DNA Plasmid Vectors Or Recombinant Baculovirus Carrying Multiple Genes For The Production Of Virus-Like Particles (VLP) Displaying Unique HA Molecules
  • FIG. 6 The general structure of the constructs generated is shown in Figure 6 .
  • the genes of interest were subcloned into mammalian plasmids (vectors, Figure 6 , panel I) containing unique regulatory sequences that enhance not only high and sustainable levels of gene expression but also the rate of generating stably transfected cells.
  • An alternative method of VLP production is the utilization of the baculovirus-insect cells expression system.
  • the overall structure of constructs generated is depicted in Figure 6 panel II.
  • influenza M1 and M2 genes were sequentially subcloned into a mammalian plasmid expression vector using the appropriate restriction sites such that each gene was under the transcriptional control of a mammalian promoters (CMV promoter or promoter A).
  • CMV promoter or promoter A a mammalian promoters
  • the plasmids into which the influenza M1 and M2 genes were cloned also contained an antibiotic selection markers (e.g., hygromicin, puromycin, or neomycin) and specific sequences upstream of each gene that maintain an open chromatin state following DNA integration within mammalian chromosomes.
  • an antibiotic selection markers e.g., hygromicin, puromycin, or neomycin
  • FIG 8A-C An example of selection of stably transfected mammalian cells using fluorescence cell sorting is shown in Figure 8A-C . Furthermore, the level of expression of M1/M2 proteins in stably transfected MDCK and CHO cells are shown in Figure 9A and B , respectively.
  • HA and NA genes were subcloned into another mammalian plasmid expression vector using the appropriate restriction sites such that each gene was under the transcriptional control of a mammalian promoters (CMV promoter or promoter A).
  • CMV promoter or promoter A a mammalian promoters
  • Plasmid DNA of each construct was further amplified by transforming MAX Efficiency Stbl2 competent E. Coli cells, and performing maxi-prep DNA preparation (Qiagen, Valencia, CA). The concentration of plasmid DNAs were determined by spectrophotometry. Transient transfections were performed-utilizing circular DNA, whereas transfections for the generation of stable cell lines were carried out with linear DNA cut with I-SceI restriction enzyme.
  • a baculovirus transfer vector was generated for VLP production in insect cells.
  • the four influenza genes were subcloned within a single plasmid and under the transcriptional control of baculovirus promoters as illustrated in Figure 6 panel II. As previously described, the genes orientation and integrity were verified by restriction enzyme analysis and sequencing. Plasmid DNA was further amplified by performing maxi-prep DNA preparation. Co-transfection of the transfer vector (carrying the four influenza genes) together with linearized baculovirus DNA into Sf9 insect cells results in the creation of recombinant baculovirus for the production of VLP vaccines.
  • the vectors were utilized for the production of VLPs in mammalian cells (CHO, Vero, MDCK, WI-38 or MRC5).
  • Cells were resuspended by adding to the flask 6ml of DMEM (Gibco) containing 10%FBS (Invitrogen, San Diego, CA) collected in a tube and subsequently pelleted by centrifugation at 500xg for 5 minutes. Cell pellet was washed twice with 5ml of ice cold 1X RPMI 1640 (Cellgro, Mediatech, Manassas, VA) and then resuspend in 500 ⁇ l of ice cold 1X RPMI.
  • DMEM Gibco
  • FBS Invitrogen, San Diego, CA
  • Cell pellet was washed twice with 5ml of ice cold 1X RPMI 1640 (Cellgro, Mediatech, Manassas, VA) and then resuspend in 500 ⁇ l of ice cold 1X RPMI.
  • the cell suspension received 6 ⁇ g of linearized plasmid expressing M1/M2, or M1/M2 plus a plasmid expressing NA/remodeled HA, gently mixed by pipeting and then transferred into a 0.4 cm gap electroporation cuvette (Bio-Rad, Hercules, CA). The cuvette was placed in a Bio-Rad Gene Pulsar, and cells electroporated using the following parameters: 400V, 960 ⁇ F. Electroporated cells were kept at room temperature for 5 minutes, then transferred into a 6-well plate in DMEM with 10% FBS and penicillin/streptomycin (Gibco) and incubated at 37°C with 5% CO2. Six hours post electroporation, the medium was aspirated, cells washed once with 1X PBS, and fresh medium added. Cells were incubated at 37°C with 5% CO 2 until antibiotic selection was initiated.
  • a modified protocol was used for the electroporation of suspension cells, e.g. CHO cell line. Cells were directly collected from the culture vessel without the need of trypsin treatment. Subsequent steps were performed as described above.
  • Mammalian cells (CHO, Vero, MDCK, MRC5 or WI-38) were prepared for transfection by plating in an appropriate culture vessel (25cm2 flasks for CHO cells or 75cm2 flasks for Vero, MDCK, MRC5 or WI-38 cells) at a density of 1.5 x 10 6 to 2.5 x 10 6 cells/ml in 5 ml of CHO-S-SFM II medium (CHO cells) or 10ml of DMEM (Vero, MDCK, MRC5, WI-38) supplemented with 5% fetal bovine serum (FBS) (Invitrogen, Carlsbad, CA). Adherent cells (Vero, MDCK, MRC5, WI-38) were plated 24 hours prior to the initiation of the transfection procedure.
  • an appropriate culture vessel 25cm2 flasks for CHO cells or 75cm2 flasks for Vero, MDCK, MRC5 or WI-38 cells
  • a DNA-lipid complexing reaction comprising of plasmid DNA with lipofectamine was set up.
  • the plasmid DNA of interest or mixture thereof was diluted in 500 ⁇ l of Opti-MEM medium in one tube and 20 ⁇ l of lipofectamine 2000 was diluted in 480 ⁇ l of Opti-MEM medium (Invitrogen, Carlsbad, CA) in another tube.
  • the lipofectamine-OptiMEM mixture was incubated at room temperature for 5 minutes.
  • the plasmid DNA-OptiMEM mixture was combined with the lipofectamine-OptiMEM mixture and the reaction was allowed to proceed at room temperature for 20 minutes.
  • the DNA-lipofectamine complex was then added to the cells previously plated as described above.
  • 2.5 ml of the contents of the 25cm2 flask was transferred to another 25cm2 flask and 4.5 ml of CHO-S -SFM II medium was added to both flasks.
  • Adherent cells were kept in the culture flask and 5ml of fresh media was added. None of the reagents or media used in the transfection process contained any antibiotics as these could get delivered to the interior of the cells by getting incorporated in the DNA-lipid complex which could prove toxic to the cells.
  • the plasmids were introduced into the cells in the form of a DNA-lipid complex in which the DNA is in its native circular form. Expression of the VLP proteins in the transfected cell lysate and in the culture supernatant was evaluated 72-96 hours post-transfection.
  • the transfection procedure was followed with the exception that the plasmid/s introduced into the cells were in a linear configuration.
  • the plasmids into which the genes of interest were cloned also contain antibiotic resistance markers e.g. Hygromycin, Puromycin or Neomycin.
  • antibiotic resistance markers e.g. Hygromycin, Puromycin or Neomycin.
  • VLPs production was performed by introducing a combination of uncut circular plasmid to mammalian cells by either electroporation or chemical transfections.
  • the M1/M2 vector was combined with a NA/HA (remodeled, FIG. 1 ) and delivered to mammalian cells (CHO, MDCK, MRC5 or WI-38) by either of the two transfection methods described.
  • a linearized M1/M2 vector ( FIG. 6A ) was introduced by electroporation or chemical transfection into MDCK, Vero or CHO cells. After this step, cells were treated with one antibiotic (hygromicin, puromycin or neomycin) chosen based on the antibiotic resistance gene carried by the plasmid.
  • the M1/M2 vector contains the hygromycin resistance gene, thus cells transfected with this plasmid were treated with this antibiotic to select stably transfected cell lines. Cells that grew in the presence of the antibiotic were tested for the expression of the M2 protein on their surface and cloned using fluorescence activated cell sorting (FACS).
  • FIG. 8 shows FACS histogram of M1/M2 transfected CHO cells which demonstrated that fraction of cell population expressed the M2 protein. Multiple cell clones were expanded and the expression of both M1 and M2 proteins further evaluated by Western blot.
  • FIG. 9A and B selected M1/M2 stably transfected MDCK and CHO cell lines constitutively express these proteins. These cell lines can be transfected with a vector carrying NA/HA (remodeled) for the assembly and release of influenza virus-like particles displaying unique HA molecules ( FIG 1-5 ).
  • Mammalian cells that had been transfected with linearized plasmids encoding gene(s) of interested were then subjected to antibiotic treatment to select out the cells in which the transfected plasmid had integrated into the host cell genome.
  • the transfected plasmid contains an antibiotic resistance cassette which confers resistance to either hygromycin, puromycin or neomycin.
  • Cells obtained as described above were harvested from the tissue culture flasks and washed three times by consecutive centrifugation at 500xg for 5 min followed by re-suspension in phosphate buffered saline (PBS). After the third wash, cells were counted and divided into three aliquots of 5x10 6 cells each. One aliquot, the one which was going to be sorted for cell surface M2 expression, was incubated with an anti-M2 monoclonal antibody (Abcam Inc, Cambridge, MA) at a dilution of 1:500 in PBS for 1 hour at room temperature.
  • an anti-M2 monoclonal antibody Abcam Inc, Cambridge, MA
  • the second aliquot was incubated with a monoclonal antibody to influenza A nucleoprotein (Meridian Life Sciences, Saco, ME) at a dilution of 1:200 in PBS for 1 hour at room temperature. This aliquot served as an isotype control for the flow cytometry experiment. The third aliquot served as the unstained control for the experiment.
  • a monoclonal antibody to influenza A nucleoprotein (Meridian Life Sciences, Saco, ME) at a dilution of 1:200 in PBS for 1 hour at room temperature.
  • This aliquot served as an isotype control for the flow cytometry experiment.
  • the third aliquot served as the unstained control for the experiment.
  • the cells were washed with PBS three times and incubated with a Fluorescein isothiocyanate (FITC) labeled anti-mouse antibody (Abcam Inc, Cambridge, MA) at a dilution of 1:100 in PBS for 1 hr at room temperature. After these treatments, the cells were washed three times with PBS and re-suspended in a final volume of 3ml of cell culture medium. These samples were then analyzed in a MoFlo cell sorter.
  • FITC Fluorescein isothiocyanate
  • FIG. 8 depicts a sorting experiment performed with M1/M2 transfected cells.
  • This selection method is applied for the identification and isolation of cells transfected with any the constructs described herein (e.g., Figures 1-5 ).
  • Cells transfected with the DNA vector carrying the NA/ HA remodeled 1 express both NA and HA proteins, which are displayed on the cell surface allowing for the identification and isolation of stably transfected clones that continuously express these proteins.
  • Selection via HA uses a broadly neutralizing monoclonal antibody that recognizes a conserved epitope on remodeled HA molecule.
  • a second transfection is performed to introduce and integrate another set of genes into the host genome of the basic cell line.
  • FIG.7B This strategy is depicted in FIG.7B , and identification and selection of stably cell lines generated on the second round of transfections is also performed by FACS analysis.
  • the five constructs of remodeled HA with or without NA which are delivered by the vector constructs depicted on FIG.6 (constructs shown in panels I) are surface molecules and displayed on the cell membrane, therefore suitable for the FACS strategy to identify and selected stably transfected cell lines that continuously produce VLPs with remodeled HA molecules.
  • Example 9 End-Point Cloning and Expansion of a Stably Transfected and Constitutively Producing M1 and M2 Influenza Proteins
  • clonal cell lines from the stably transfected cell population, end point cloning was performed.
  • the cells were harvested and counted using a hemocytometer.
  • the cells were then diluted in an appropriate volume of culture medium such that the final concentration reached 10 cells per ml.
  • This cell preparation was gently agitated to ensure homogenous cell distribution and then plated into sterile 96 well plates at 100 ⁇ l/well.
  • the plates were then incubated at 37°C with a humidified atmosphere of 5% CO2 and monitored regularly for clonal cell growth.
  • Wells with actively growing cells were identified over a period of time. When the clonal cells in these wells reached about 70-80% confluency they were scaled up by sequential passages to 6 well plates, 25 cm 2 and 75 cm 2 flasks.
  • the total protein concentration in each sample was estimated by the Bradford method; briefly, 10 ⁇ l of the sample is added to 1.0 ml of 1x Bradford Dye reagent (Bio-Rad Inc., Hercules, CA) which was prewarmed to room temperature. This reaction mixture is then shaken vigorously to create a homogenous solution. The absorbance of each sample was measured in a spectrophotometer at a wavelength of 595 nm. The protein concentration of each sample was determined using a standard curve which was plotted by measuring absorbance at 595 nm of known concentrations of bovine serum albumin using the Bradford assay.
  • Example 11 Selection of a Cell Line that Continuously Expresses the Influenza M1 and M2 Proteins
  • MDCK and CHO cells were transfected by electroporation with a linearized DNA vector carrying the M1 and M2 influenza genes. Following selection with hygromycin, single cell clones were isolated using fluorescence-activated cell sorting (FACS) after labeling the cells with combination of antibodies; first as primary an anti-M2 mouse monoclonal antibody which reacted the M2 protein expressed on the cell surface, followed by a flourescein conjugated anti-mouse as secondary.
  • FACS fluorescence-activated cell sorting
  • Sorted single cells were expanded and expression of M1 and M2 proteins was further assessed by Western blot.
  • Cells were lysed, loaded onto and SDS-PAGE (10-20%) and separated by electrophoresis. The proteins were transferred to a PVDF membrane and blocked with 4% skim milk for 1 hour. The membrane was then incubated overnight with a mouse monoclonal anti-M1 protein (1: 40,000 dilution) and a mouse monoclonal anti-M2 protein (1: 1000 dilution) (Abcam, clone 14C2,).
  • the membrane was washed 3X for 5 minutes with 1X TBST and then incubated for 1 hour with horseradish peroxidase conjugated goat anti-mouse IgG (1: 50,000 dilution in 2% skim milk) (Thermo Scientific, Rockford, IL).
  • the membrane was washed 3X for 10 minutes with 1X TBST followed by 5 minute incubation with SuperSignal West Pico chemiluminescent substrate (Thermo Scientific, Rockford, IL).
  • the membrane was exposed to HyBlot CL autoradiography film (Denville Scientific, Metuchen, NJ).
  • the MDCK cell lysate produced two major bands which correspond to the M1 protein molecular weight (MW) of ⁇ 27kDA and M2 protein with a MW of ⁇ 1kDA, whereas untransfected control did not show the presence of these proteins.
  • Lane 1 protein marker
  • lane 2 MDCK cells not expressing influenza M1 and M2 proteins (negative control)
  • lane 3 MDCK cells constitutively expressing influenza A M1 and M2 proteins. Similar results were obtained with the same plasmid was introduced into CHO cells as shown in FIG. 9B and VERO cells.
  • Example 12 Expression of Remodeled HA/NA Protein in M1/M2 Producing Cells
  • FIG. 10 Western blot analysis of these samples are shown in FIG. 10 and show production of M1, M2 and HA protein in the transfected cells.
  • Example 13 Examination of influenza VLP by Negative Staining Electron Microscopy
  • the supernatant of the transfected cells was subjected to ultra-centrifugation at 200,000xg for 1.5 hours at 4°C.
  • the pellet from the ultra-centrifugation was re-suspended in PBS and 20 ⁇ l of the re-suspended pellet was set aside for electron microscopy.
  • This 20 ⁇ l sample was fixed with 4 % paraformaldehyde and then 5 ⁇ l of the fixed material was applied to a 200 mesh carbon coated grid (EMS, Hatfield, PA) and allowed to cover the grid for 5 minutes and then washed with water three times. Following this the coated grids were exposed to five drops of 2 % uranyl acetate in quick succession to prevent over-staining of the grid with uranyl acetate.
  • Excessive solution was blotted from the grid and then air dried before loading it onto a JOEL JEM 100CX Transmission Electron Microscope. The samples were observed at a magnification of 60,000X to 100,000X.
  • FIG. 11 shows images obtained from purified supernatant of M1/M2-HA/NA CHO cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Claims (14)

  1. Particule de type virus comprenant :
    au moins une protéine de matrice ; et
    un polypeptide d'hémagglutinine d'influenza modifié comprenant un fragment d'hémagglutinine 1 et un fragment d'hémagglutinine 2 liés opérationnellement l'un à l'autre, où la modification comprend une délétion d'au moins 150 résidus d'acides aminés d'un polypeptide d'hémagglutinine 1, où le fragment d'hémagglutinine 2 comprend le domaine transmembranaire et de queue cytoplasmique du polypeptide d'hémagglutinine
    et
    où la modification comprend des mutations dans un ou plusieurs acides aminés d'un polypeptide d'hémagglutinine 2.
  2. Particule de type virus selon la revendication 1, dans laquelle au moins un site de clivage protéolytique ou au moins un lieur est inséré dans le polypeptide d'hémagglutinine d'influenza modifié.
  3. Particule de type virus selon les revendications 1 ou 2, dans laquelle le domaine transmembranaire du polypeptide d'hémagglutinine d'influenza modifié est remplacé par un domaine transmembranaire d'hémagglutinine d'une souche ou d'un sous-type différent(e) du polypeptide d'hémagglutinine d'influenza modifié et/ou où le domaine cytoplasmique du polypeptide d'hémagglutinine d'influenza modifié est remplacé par un domaine cytoplasmique d'une souche ou d'un sous-type différent(e) du polypeptide d'hémagglutinine d'influenza modifié.
  4. Particule de type virus selon l'une quelconque des revendications 1 à 3, comprenant en outre des protéines d'influenza supplémentaires telles que une ou plusieurs protéines de matrice sauvages, une ou plusieurs protéines de matrice mutantes, une ou plusieurs protéines de matrice hybrides, une ou plusieurs protéines de matrice mutantes et hybrides, une ou plusieurs glycoprotéines antigéniques sauvages, une ou plusieurs glycoprotéines antigéniques modifiées, une ou plusieurs glycoprotéines antigéniques hybrides, une ou plusieurs glycoprotéines antigéniques modifiées et hybrides, une ou plusieurs nucléoprotéines, une ou plusieurs protéines PB1, une ou plusieurs protéines PB2, une ou plusieurs protéines PA ou leurs combinaisons.
  5. Particule de type virus selon l'une quelconque des revendications 1 à 4, où la particule de type virus est exprimée dans une cellule eucaryote dans des conditions qui permettent l'assemblage et la libération de particules de type virus.
  6. Particule de type virus selon la revendication 5, dans laquelle la cellule eucaryote est choisie dans le groupe constitué par une cellule de levure, une cellule d'insecte, une cellule amphibienne, une cellule aviaire, une cellule végétale ou une cellule mammifère.
  7. Procédé de production d'une particule de type virus selon l'une quelconque des revendications 1 à 6, le procédé comprenant les étapes de :
    transfection d'un ou de plusieurs vecteurs codant pour au moins une protéine de matrice et au moins un polypeptide d'hémagglutinine d'influenza modifié comprenant un fragment d'hémagglutinine 1 et un fragment d'hémagglutinine 2 liés opérationnellement l'un à l'autre, dans une cellule hôte appropriée, où la modification comprend une délétion d'au moins 150 résidus d'acide aminé d'un polypeptide d'hémagglutinine 1, où le fragment d'hémagglutinine 2 comprend le domaine transmembranaire et de queue cytoplasmique du polypeptide d'hémagglutinine,
    et où la modification comprend des mutations dans un ou plusieurs acides aminés d'un polypeptide d'hémagglutinine 2,
    et l'expression de la combinaison de protéines dans des conditions qui permettent la formation de particules de type virus.
  8. Procédé selon la revendication 7, dans lequel au moins une protéine de matrice est choisie dans le groupe constitué par une protéine M1 d'influenza, une protéine de matrice thogoto et une protéine de matrice RSV et en outre où au moins un vecteur comprend en outre éventuellement une séquence codant pour une protéine M2 d'influenza.
  9. Procédé selon la revendication 7 ou la revendication 8, dans lequel les un ou plusieurs vecteurs sont transfectés de manière stable dans la cellule hôte et éventuellement où l'au moins une protéine de matrice et le polypeptide d'hémagglutinine d'influenza modifié sont codés sur des vecteurs séparés et en outre où le vecteur codant pour l'au moins une protéine de matrice est transfecté de manière stable dans la cellule avant la transfection avec le vecteur codant pour la protéine de polypeptide d'hémagglutinine d'influenza modifié.
  10. Composition immunogène comprenant au moins une particule de type virus selon l'une quelconque des revendications 1 à 6 et comprenant en outre un adjuvant.
  11. Composition immunogène selon la revendication 10, où la composition comprend au moins deux particules de type virus comprenant des polypeptides d'hémagglutinine d'influenza modifiés différents.
  12. Particule de type virus selon l'une quelconque des revendications 1 à 6 ou composition immunogène selon la revendication 10 ou 11 pour une utilisation en thérapie ou prévention dans un procédé de production d'une réponse immunitaire à l'influenza chez un sujet, le procédé comprenant l'administration au sujet d'une quantité efficace de ladite particule de type virus ou de ladite composition immunogène.
  13. Particule de type virus selon l'une quelconque des revendications 1 à 6 ou composition immunogène selon la revendication 10 ou 11 pour une utilisation dans un procédé selon la revendication 12, où la composition est administrée par voie mucosale, intradermique, sous-cutanée, intramusculaire ou orale.
  14. Particule de type virus selon l'une quelconque des revendications 1 à 6 ou composition immunogène selon la revendication 10 ou 11 pour une utilisation dans un procédé selon la revendication 12 ou 13, où la réponse immunitaire vaccine le sujet contre de multiples souches ou sous-types d'influenza.
EP11744998.3A 2010-02-18 2011-02-18 Vaccins universels contre la grippe à pseudo-particules virales (vlp) Not-in-force EP2536428B1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US30576810P 2010-02-18 2010-02-18
US30575910P 2010-02-18 2010-02-18
PCT/US2011/000300 WO2011102900A1 (fr) 2010-02-18 2011-02-18 Vaccins universels contre la grippe à pseudo-particules virales (vlp)

Publications (3)

Publication Number Publication Date
EP2536428A1 EP2536428A1 (fr) 2012-12-26
EP2536428A4 EP2536428A4 (fr) 2014-04-09
EP2536428B1 true EP2536428B1 (fr) 2018-12-05

Family

ID=44483229

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11744998.3A Not-in-force EP2536428B1 (fr) 2010-02-18 2011-02-18 Vaccins universels contre la grippe à pseudo-particules virales (vlp)

Country Status (6)

Country Link
US (4) US9352031B2 (fr)
EP (1) EP2536428B1 (fr)
JP (2) JP2013520167A (fr)
CN (1) CN102858368B (fr)
CA (1) CA2789945A1 (fr)
WO (1) WO2011102900A1 (fr)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2615372A1 (fr) 2007-07-13 2009-01-13 Marc-Andre D'aoust Particules semblables au virus grippal a comprenant de l'hemagglutinine
PT2610345E (pt) 2007-11-27 2016-01-11 Medicago Inc Partículas semelhantes ao vírus (vlps) da gripe recombinantes produzidas em plantas transgénicas que expressam hemaglutinina
WO2011102900A1 (fr) * 2010-02-18 2011-08-25 Technovax, Inc. Vaccins universels contre la grippe à pseudo-particules virales (vlp)
WO2012024283A2 (fr) * 2010-08-16 2012-02-23 The Wistar Institute Of Anatomy And Biology Vaccins universels contre la grippe a
EP2758038B1 (fr) * 2011-09-23 2018-05-30 The United States of America, as represented by The Secretary, Department of Health & Human Services Nouveaux vaccins à base de protéine hémagglutinine de la grippe
PT2760882T (pt) 2011-09-30 2023-08-07 Medicago Inc Aumento do rendimento de partícula semelhante a vírus em plantas
US11390878B2 (en) 2011-09-30 2022-07-19 Medicago Inc. Increasing protein yield in plants
WO2013059403A1 (fr) * 2011-10-19 2013-04-25 Immunotope, Inc. Immunogènes induisant des lymphocytes t cytotoxiques pour prévenir, traiter, et diagnostiquer l'infection par le virus de l'influenza
US9364530B2 (en) 2012-03-22 2016-06-14 Fraunhofer Usa, Inc. Virus-like particles comprising a matrix protein from a plant enveloped virus and uses thereof
EP2911690A1 (fr) 2012-10-26 2015-09-02 MannKind Corporation Compositions et procédés de vaccin antigrippal inhalable
EP3626827A1 (fr) 2013-03-28 2020-03-25 Medicago Inc. Production de particules de type viral de la grippe dans des plantes
CN105848722B (zh) * 2013-10-02 2021-09-03 免疫医疗有限责任公司 中和抗甲型流感抗体及其用途
BR112016007868A2 (pt) 2013-10-11 2017-12-05 Us Health vacinas contra o vírus de epstein-barr
WO2015101666A1 (fr) 2014-01-03 2015-07-09 Fundación Biofísica Bizkaia Vlp, procédés pour leur obtention et applications de ceux-ci
WO2015195218A1 (fr) * 2014-06-20 2015-12-23 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Particules pseudo-virales (ppv) grippales polyvalentes, et leur utilisation comme vaccins
CA2974346A1 (fr) 2014-12-31 2016-07-07 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Nouveaux vaccins multivalents a base de nanoparticules
CN108348593B (zh) * 2015-08-31 2022-08-16 泰克诺瓦克斯股份有限公司 基于人呼吸道合胞病毒(hrsv)病毒样颗粒(vlps)的疫苗
AU2017321883B2 (en) 2016-09-02 2022-07-07 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Stabilized group 2 influenza hemagglutinin stem region trimers and uses thereof
MX2019014943A (es) * 2018-12-12 2020-08-06 Cambridge Tech Llc Vacuna universal contra la gripe.
CN110272473B (zh) * 2019-07-10 2021-03-02 军事科学院军事医学研究院军事兽医研究所 甲型流感通用型病毒样颗粒及其制备方法和应用
EP4196160A2 (fr) * 2020-08-17 2023-06-21 Technovax, Inc. Vaccin à particule de type virus [vlp] du coronavirus à syndrome respiratoire aigu sévère [sars-cov-2] : compositions, stratégies d'administration, méthodes et utilisations
CN112342149A (zh) * 2020-11-12 2021-02-09 新疆畜牧科学院兽医研究所(新疆畜牧科学院动物临床医学研究中心) 一种禽流感病毒样颗粒组装表达系统及其应用
WO2023126982A1 (fr) * 2021-12-31 2023-07-06 Mynvax Private Limited Fragments polypeptidiques, composition immunogène contre le virus de la grippe et mises en œuvre associées

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2410297C (fr) 2000-06-23 2011-03-15 American Cyanamid Company Assemblage sauvage-chimerique de particules viraloides de la grippe (vlp)
US8592197B2 (en) * 2003-07-11 2013-11-26 Novavax, Inc. Functional influenza virus-like particles (VLPs)
US8080255B2 (en) * 2003-07-11 2011-12-20 Novavax Inc. Functional influenza virus like particles (VLPs)
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
MX2008000890A (es) * 2005-07-19 2008-03-18 Dow Global Technologies Inc Vacunas de influenza (flu) recombinantes.
TWI488968B (zh) * 2005-10-18 2015-06-21 Novavax Inc 功能性流行性感冒類病毒顆粒(VLPs)
US9101578B2 (en) 2006-05-01 2015-08-11 Technovax, Inc. Polyvalent influenza virus-like particle (VLP) compositions
WO2007130327A2 (fr) 2006-05-01 2007-11-15 Technovax, Inc. Compositions de particules de type viral (vlp) de la grippe
EP2044224A4 (fr) * 2006-07-27 2011-04-13 Ligocyte Pharmaceuticals Inc Particules de type virus chimérique
WO2008061243A2 (fr) 2006-11-16 2008-05-22 Novavax, Inc. Particules de type viral (vlp) apparentées au virus respiratoire syncytial
JP5762969B2 (ja) * 2008-11-25 2015-08-12 バクスター・ヘルスケヤー・ソシエテ・アノニムBaxter Healthcare SA pH安定性のエンベロープ付きウィルスの生産方法
AU2010234849B2 (en) * 2009-03-30 2017-06-22 Mount Sinai School Of Medicine Influenza virus vaccines and uses thereof
WO2011102900A1 (fr) * 2010-02-18 2011-08-25 Technovax, Inc. Vaccins universels contre la grippe à pseudo-particules virales (vlp)
KR102072582B1 (ko) * 2012-12-31 2020-02-03 엘지전자 주식회사 듀얼 디스플레이 방법 및 장치

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
JP6524555B2 (ja) 2019-06-05
EP2536428A1 (fr) 2012-12-26
JP2013520167A (ja) 2013-06-06
CN102858368A (zh) 2013-01-02
US10080796B2 (en) 2018-09-25
US9352031B2 (en) 2016-05-31
WO2011102900A1 (fr) 2011-08-25
US20110212128A1 (en) 2011-09-01
US10695418B2 (en) 2020-06-30
CA2789945A1 (fr) 2011-08-25
US20190091323A1 (en) 2019-03-28
EP2536428A4 (fr) 2014-04-09
US20160303223A1 (en) 2016-10-20
JP2016198107A (ja) 2016-12-01
US20200297836A1 (en) 2020-09-24
US11529410B2 (en) 2022-12-20
CN102858368B (zh) 2017-04-12

Similar Documents

Publication Publication Date Title
US11529410B2 (en) Universal virus-like particle (VLP) influenza vaccines
US9387241B2 (en) Influenza virus-like particle (VLP) compositions
US9101578B2 (en) Polyvalent influenza virus-like particle (VLP) compositions
US20110097358A1 (en) RESPIRATORY SYNCYTIAL VIRUS (RSV) VIRUS-LIKE PARTICLES (VLPs)
JP6643239B2 (ja) 免疫原性の中東呼吸器症候群コロナウイルス(MERS−CoV)組成物および方法
US7507411B2 (en) Attenuated influenza NS1 variants
AU2004249133B2 (en) High titer recombinant influenza viruses for vaccines and gene therapy
US9884105B2 (en) Production of viral vaccine
EP1339738B1 (fr) Polynucleotides codant pour des polypeptides antigeniques du type c du vih, de tels polypeptides et leurs utilisations
US20080089908A1 (en) Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20190030156A1 (en) Human respiratory syncytial virus (hrsv) virus-like particles (vlps) based vaccine
AU2008275513A1 (en) Enhancement of glycoprotein incorporation into virus-like particles
EP2292772A1 (fr) Vaccination VIH avec un ADN codant un polypeptide VIH et un polypeptide VIH
KR101835989B1 (ko) 인플루엔자 바이러스의 다중 아형 h3 및 h7 에 대한 다중 교차 면역반응을 형성하는 신규한 재조합 인플루엔자 바이러스 및 이를 포함하는 백신
US20130189303A1 (en) Recombinant swine influenza virus and uses thereof
CN1644686B (zh) 流感病毒哺乳动物细胞高产毒株、其重组毒株及其制备方法和应用

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120918

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20140307

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 15/09 20060101ALI20140304BHEP

Ipc: A61K 39/145 20060101AFI20140304BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170223

INTG Intention to grant announced

Effective date: 20180611

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAR Information related to intention to grant a patent recorded

Free format text: ORIGINAL CODE: EPIDOSNIGR71

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

INTC Intention to grant announced (deleted)
GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

INTG Intention to grant announced

Effective date: 20181026

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1072229

Country of ref document: AT

Kind code of ref document: T

Effective date: 20181215

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011054522

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20181205

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1072229

Country of ref document: AT

Kind code of ref document: T

Effective date: 20181205

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190305

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190306

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190405

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190405

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602011054522

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190218

26N No opposition filed

Effective date: 20190906

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20190228

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20190305

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190218

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190903

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190305

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20110218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181205